PPAR[gamma]-coactivator-1[alpha] gene transfer reduces neuronal loss and amyloid-[Beta] generation by reducing [Beta]-secretase in an Alzheimer's disease model

Current therapies for Alzheimer's disease (AD) are symptomatic and do not target the underlying A[beta] pathology and other important hallmarks including neuronal loss. PPAR[gamma]-coactivator-1[alpha] (PGC-1[alpha]) is a cofactor for transcription factors including the peroxisome proliferator-...

Full description

Saved in:
Bibliographic Details
Published inProceedings of the National Academy of Sciences - PNAS Vol. 113; no. 43; p. 12292
Main Authors Katsouri, Loukia, Lim, Yau M, Blondrath, Katrin, Eleftheriadou, Ioanna, Lombardero, Laura, Birch, Amy M, Mirzaei, Nazanin, Irvine, Elaine E, Mazarakis, Nicholas D, Sastre, Magdalena
Format Journal Article
LanguageEnglish
Published Washington National Academy of Sciences 25.10.2016
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Current therapies for Alzheimer's disease (AD) are symptomatic and do not target the underlying A[beta] pathology and other important hallmarks including neuronal loss. PPAR[gamma]-coactivator-1[alpha] (PGC-1[alpha]) is a cofactor for transcription factors including the peroxisome proliferator-activated receptor-[gamma] (PPAR[gamma]), and it is involved in the regulation of metabolic genes, oxidative phosphorylation, and mitochondrial biogenesis. We previously reported that PGC-1[alpha] also regulates the transcription of [beta]-APP cleaving enzyme (BACE1), the main enzyme involved in A[beta] generation, and its expression is decreased in AD patients. We aimed to explore the potential therapeutic effect of PGC-1[alpha] by generating a lentiviral vector to express human PGC-1[alpha] and target it by stereotaxic delivery to hippocampus and cortex of APP23 transgenic mice at the preclinical stage of the disease. Four months after injection, APP23 mice treated with hPGC-1[alpha] showed improved spatial and recognition memory concomitant with a significant reduction in A[beta] deposition, associated with a decrease in BACE1 expression. hPGC-1[alpha] overexpression attenuated the levels of proinflammatory cytokines and microglial activation. This effect was accompanied by a marked preservation of pyramidal neurons in the CA3 area and increased expression of neurotrophic factors. The neuroprotective effects were secondary to a reduction in A[beta] pathology and neuroinflammation, because wild-type mice receiving the same treatment were unaffected. These results suggest that the selective induction of PGC-1[alpha] gene in specific areas of the brain is effective in targeting AD-related neurodegeneration and holds potential as therapeutic intervention for this disease.
ISSN:0027-8424
1091-6490